Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Vet Microbiol ; 254: 109000, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33515926

ABSTRACT

The first avian H7N9 influenza outbreak in spring of 2013 emerged in an unprecedented transmission from infected poultry to humans in the Yangtze delta area, eastern China, posing a dual challenge to public health and poultry industry. However, the mechanism for how avian H7N9 influenza virus adapts to mammalian hosts has not been clearly understood. Here, to identify adaptive changes that confer enhanced virulence of H7N9 virus in mammals, we generated a mouse-adapted H7N9 variant virus (S8) by serial lung-to-lung passages of the wild-type SDL124 virus in mice and compared their phenotype in vivo and in vitro. Sequence analysis showed that the two viruses differed by 27 amino acids distributed among six genes, containing changes in PB2 (E627K, D701N) and HA (Q226L) genes. The 50% mouse lethal dose (MLD50) of S8 reduced about 500 folds, to be moderately pathogenic to mice when compared to that of low pathogenic wild-type SDL124. Moreover, S8 replicated efficiently in mouse lungs and displayed expanded tissue tropism, and induced a greater degree of pulmonary edema and higher level of inflammatory cell infiltration in bronchoalveolar lavage fluids than SDL124 did. Interestingly, the mouse adapted S8 virus obtained strong affinity for human-like (SAα-2,6 Gal) receptor during the adaptation in mice. Correspondingly, compared with SDL124 virus, S8 virus showed higher replication efficiency in mammalian cells, whereas lower replication ability in avian cells. Taken together, these findings suggest that these mutations synergistically elevate the ability of H7N9 virus to disseminate to multiple organs and subsequently enhance the virulence of H7N9 virus in mammalian hosts.


Subject(s)
Adaptation, Physiological/genetics , Influenza A Virus, H7N9 Subtype/genetics , Influenza A Virus, H7N9 Subtype/pathogenicity , Lung/microbiology , Mutation , Sialic Acids/metabolism , A549 Cells , Animals , Dogs , Female , Humans , Lung Injury/virology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Sequence Analysis, DNA , Serial Passage , Viral Proteins/genetics , Viral Tropism , Virulence/genetics , Virus Replication
2.
Arch Virol ; 162(11): 3517-3522, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28744591

ABSTRACT

Nuclear factor kappa B (NF-κB) plays a crucial role in inflammation and immune responses. Our previous studies have demonstrated that the innate immune response affect H5N1 virus virulence in mice. In this study, we first showed that the PA protein of the highly pathogenic avian influenza virus strain CK10 had the strongest inhibitory effect on NF-κB activation when compared with other genes, and that it acted in a dose independent-manner. We then determined the critical amino acids of PA that contribute to this effect. Furthermore, PA also inhibited NF-κB-regulated inflammatory factors, including IL-6, IL-2, Nos-2 and TNF-α. However, the inhibitory effect on NF-κB activation mediated by PA was not associated with nuclear translocation of p65.


Subject(s)
Influenza A Virus, H5N1 Subtype/metabolism , NF-kappa B/metabolism , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/metabolism , Virulence Factors/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Influenza A Virus, H5N1 Subtype/genetics , RNA-Dependent RNA Polymerase/genetics , Viral Proteins/genetics , Virulence Factors/genetics
3.
Front Microbiol ; 8: 739, 2017.
Article in English | MEDLINE | ID: mdl-28503168

ABSTRACT

Accumulating data have identified the important roles of PA protein in replication and pathogenicity of influenza A virus (IAV). Identification of host factors that interact with the PA protein may accelerate our understanding of IAV pathogenesis. In this study, using immunoprecipitation assay combined with liquid chromatography-tandem mass spectrometry, we identified 278 human cellular proteins that might interact with PA of H5N1 IAV. Gene Ontology annotation revealed that the identified proteins are highly associated with viral translation and replication. Further KEGG pathway analysis of the interactome profile highlighted cellular pathways associated with translation, infectious disease, and signal transduction. In addition, Diseases and Functions analysis suggested that these cellular proteins are highly related with Organismal Injury and Abnormalities and Cell Death and Survival. Moreover, two cellular proteins (nucleolin and eukaryotic translation elongation factor 1-alpha 1) identified both in this study and others were further validated to interact with PA using co-immunoprecipitation and co-localization assays. Therefore, this study presented the interactome data of H5N1 IAV PA protein in human cells which may provide novel cellular target proteins for elucidating the potential molecular functions of PA in regulating the lifecycle of IAV in human cells.

4.
Med Microbiol Immunol ; 206(2): 125-147, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28000052

ABSTRACT

We previously reported a pair of H5N1 avian influenza viruses which are genetically similar but differ greatly in their virulence in mice. A/Chicken/Jiangsu/k0402/2010 (CK10) is highly lethal to mice, whereas A/Goose/Jiangsu/k0403/2010 (GS10) is avirulent. In this study, to investigate the host factors that account for their virulence discrepancy, we compared the pathology and host proteome of the CK10- or GS10-infected mouse lung. Moderate lung injury was observed from CK10-infected animals as early as the first day of infection, and the pathology steadily progressed at later time point. However, only mild lesions were observed in GS10-infected mouse lung at the late infection stage. Using the quantitative iTRAQ coupled LC-MS/MS method, we first found that more significantly differentially expressed (DE) proteins were stimulated by GS10 compared with CK10. However, bio-function analysis of the DE proteins suggested that CK10 induced much stronger inflammatory response-related functions than GS10. Canonical pathway analysis also demonstrated that CK10 highly activated the "Acute Phase Response Signaling," which results in a wide range of biological activities in response to viral infection, including many inflammatory processes. Further in-depth analysis showed that CK10 exacerbated acute lung injury-associated responses, including inflammatory response, cell death, reactive oxygen species production and complement response. In addition, some of these identified proteins that associated with the lung injury were further confirmed to be regulated in vitro. Therefore, our findings suggest that the early increased lung injury-associated host response induced by CK10 may contribute to the lung pathology and the high virulence of this virus in mice.


Subject(s)
Host-Pathogen Interactions , Influenza A Virus, H5N1 Subtype , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Proteome , Proteomics/methods , Acute-Phase Reaction/metabolism , Acute-Phase Reaction/virology , Animals , Cell Communication , Cell Line , Chromatography, Liquid , Cluster Analysis , Computational Biology/methods , Female , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Influenza A Virus, H5N1 Subtype/pathogenicity , Mice , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/pathology , Protein Interaction Mapping , Protein Interaction Maps , Reproducibility of Results , Signal Transduction , Tandem Mass Spectrometry
5.
Vet Microbiol ; 192: 95-101, 2016 Aug 30.
Article in English | MEDLINE | ID: mdl-27527770

ABSTRACT

Reassortment between different influenza viruses is a crucial way to generate novel influenza viruses with unpredictable virulence and transmissibility, which may threaten the public health. As currently in China, avian influenza viruses (AIVs) of H9N2 and H5N1 subtypes are endemic in poultry in many areas, while they are prone to reassort with each other naturally. In order to evaluate the risk of the reassortment to public health, A/Goose/Jiangsu/k0403/2010 [GS/10(H5N1)] virus was used as a backbone to generate a series of reassortants, each contained a single internal gene derived from the predominant S genotype of the A/Chicken/Jiangsu/WJ57/2012 [WJ/57(H9N2)]. We next assessed the biological characteristics of these assortments, including pathogenicity, replication efficiency and polymerase activity. We found that the parental WJ/57(H9N2) and GS/10(H5N1) viruses displayed high genetic compatibility. Notably, the H5N1 reassortants containing the PA or NP gene from WJ/57(H9N2) virus significantly increased virulence and replication ability in mice, as well as markedly enhanced polymerase activity. Our results indicate that the endemicity of H9N2 and H5N1 in domestic poultry greatly increases the possibility of generating new viruses by reassortment that may pose a great threat to poultry industry and public health.


Subject(s)
Genes, Viral/genetics , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H9N2 Subtype/genetics , Orthomyxoviridae Infections/virology , Reassortant Viruses/genetics , Animals , Cell Line , Dogs , Humans , Influenza A Virus, H5N1 Subtype/pathogenicity , Mice , Viral Tropism , Virulence
6.
Med Microbiol Immunol ; 205(4): 381-95, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27289459

ABSTRACT

PA-X is a novel discovered accessory protein encoded by the PA mRNA. Our previous study demonstrated that PA-X decreases the virulence of a highly pathogenic H5N1 strain A/Chicken/Jiangsu/k0402/2010 in mice. However, the underlying mechanism of virulence attenuation associated with PA-X is still unknown. In this study, we compared two PA-X-deficient mutant viruses and the parental virus in terms of induction of pathology and manipulation of host response in the mouse lung, stimulation of cell death and PA nuclear accumulation. We first found that down-regulated PA-X expression markedly aggravated the acute lung injury of the infected mice early on day 1 post-infection (p.i.). We then determined that loss of PA-X expression induced higher levels of cytokines, chemokines and complement-derived peptides (C3a and C5a) in the lung, especially at early time point's p.i. In addition, in vitro assays showed that the PA-X-deficient viruses enhanced cell death and increased expression of reactive oxygen species (ROS) in mammalian cells. Moreover, we also found that PA nuclear accumulation of the PA-X-null viruses accelerated in MDCK cells. These results demonstrate that PA-X decreases the level of complement components, ROS, cell death and inflammatory response, which may together contribute to the alleviated lung injury and the attenuation of the virulence of H5N1 virus in mice.


Subject(s)
Acute Lung Injury/pathology , Acute Lung Injury/virology , Influenza A Virus, H5N1 Subtype/growth & development , Influenza A Virus, H5N1 Subtype/immunology , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Repressor Proteins/immunology , Viral Nonstructural Proteins/immunology , Animals , Cell Death , Complement System Proteins/analysis , Cytokines/analysis , Disease Models, Animal , Dogs , Female , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/pathogenicity , Lung/pathology , Madin Darby Canine Kidney Cells , Mice, Inbred BALB C , Reactive Oxygen Species/analysis , Repressor Proteins/deficiency , Viral Nonstructural Proteins/deficiency
7.
Virus Res ; 210: 255-63, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26315686

ABSTRACT

The highly pathogenic A/chicken/Hebei/1102/2010 (HB10) H5N2 virus is a natural reassortant derived from circulating H5N1 and endemic H9N2 avian influenza viruses (AIV). To evaluate the potential of its interspecies transmission, we previously serially passaged the non-virulent HB10 virus in the mouse lung and obtained a high virulence variant (HB10-MA). Genomic sequencing revealed five mutations (HA-S227N, PB2-Q591K, PB2-D701N, PA-I554V and NP-R351K) that distinguished HB10-MA virus from its parental HB10 virus. In this study, we further investigated the molecular basis for the enhanced virulence of HB10-MA in mice. By generating a series of reassortants between the two viruses and evaluating their virulence in mice, we found that both PB2 and PA genes contribute to the high virulence of HB10-MA in mice, whereas PB2 gene carrying the 591K and/or 701N had a dominant function. In addition, the two amino acids showed a cumulative effect on the virulence, virus replication, and polymerase activity of HB10 or HB10-MA. Therefore, our results collectively emphasized the crucial role of PB2 gene, particularly the paired mutations of Q591K and D701N in the host adaptation of the novel reassortant H5N2 AIV in mammals, which may provide helpful insights into the pathogenic potential of emerging AIV in human beings.


Subject(s)
Adaptation, Biological , Influenza A Virus, H5N2 Subtype/genetics , Influenza A Virus, H5N2 Subtype/pathogenicity , Mutation, Missense , RNA-Dependent RNA Polymerase/genetics , Reassortant Viruses/genetics , Reassortant Viruses/pathogenicity , Viral Proteins/genetics , Animals , Chickens , Female , Genome, Viral , Influenza A Virus, H5N2 Subtype/enzymology , Influenza A Virus, H5N2 Subtype/growth & development , Influenza in Birds/virology , Mice, Inbred BALB C , Orthomyxoviridae Infections/virology , RNA-Dependent RNA Polymerase/metabolism , Reassortant Viruses/enzymology , Reverse Genetics , Sequence Analysis, DNA , Serial Passage , Viral Proteins/metabolism , Virulence , Virulence Factors/genetics , Virulence Factors/metabolism
8.
J Virol ; 89(8): 4126-42, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25631083

ABSTRACT

UNLABELLED: PA-X is a newly discovered protein that decreases the virulence of the 1918 H1N1 virus in a mouse model. However, the role of PA-X in the pathogenesis of highly pathogenic avian influenza viruses (HPAIV) of the H5N1 subtype in avian species is totally unknown. By generating two PA-X-deficient viruses and evaluating their virulence in different animal models, we show here that PA-X diminishes the virulence of the HPAIV H5N1 strain A/Chicken/Jiangsu/k0402/2010 (CK10) in mice, chickens, and ducks. Expression of PA-X dampens polymerase activity and virus replication both in vitro and in vivo. Using microarray analysis, we found that PA-X blunts the global host response in chicken lungs, markedly downregulating genes associated with the inflammatory and cell death responses. Correspondingly, a decreased cytokine response was recapitulated in multiple organs of chickens and ducks infected with the wild-type virus relative to those infected with the PA-X-deficient virus. In addition, the PA-X protein exhibits antiapoptotic activity in chicken and duck embryo fibroblasts. Thus, our results demonstrated that PA-X acts as a negative virulence regulator and decreases virulence by inhibiting viral replication and the host innate immune response. Therefore, we here define the role of PA-X in the pathogenicity of H5N1 HPAIV, furthering our understanding of the intricate pathogenesis of influenza A virus. IMPORTANCE: Influenza A virus (IAV) continues to pose a huge threat to global public health. Eight gene segments of the IAV genome encode as many as 17 proteins, including 8 main viral proteins and 9 accessory proteins. The presence of these accessory proteins may further complicate the pathogenesis of IAV. PA-X is a newly identified protein in segment 3 that acts to decrease the virulence of the 1918 H1N1 virus in mice by modulating host gene expression. Our study extends these functions of PA-X to H5N1 HPAIV. We demonstrated that loss of PA-X expression increases the virulence and replication of an H5N1 virus in mice and avian species and alters the host innate immune and cell death responses. Our report is the first to delineate the role of the novel PA-X protein in the pathogenesis of H5N1 viruses in avian species and promotes our understanding of H5N1 HPAIV.


Subject(s)
Chickens , Host-Pathogen Interactions/genetics , Influenza A Virus, H5N1 Subtype/metabolism , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza in Birds/virology , Repressor Proteins/metabolism , Viral Nonstructural Proteins/metabolism , Virus Replication/genetics , Animals , Base Sequence , Blotting, Western , Cell Fractionation , Cell Line , Dogs , Ducks , Fluorescent Antibody Technique , Humans , Influenza in Birds/metabolism , Luciferases , Mice , Microarray Analysis , Molecular Sequence Data , Mutation/genetics , Repressor Proteins/genetics , Sequence Analysis, DNA , Viral Nonstructural Proteins/genetics
9.
J Gen Virol ; 95(Pt 5): 1067-1073, 2014 May.
Article in English | MEDLINE | ID: mdl-24603525

ABSTRACT

The Newcastle disease virus (NDV) matrix (M) protein is a highly basic and nucleocytoplasmic shuttling viral protein. Previous study has demonstrated that the N-terminal 100 aa of NDV M protein are somewhat acidic overall, but the remainder of the polypeptide is strongly basic. In this study, we investigated the role of the N-terminal basic residues in the subcellular localization of M protein and in the replication and pathogenicity of NDV. We found that mutation of the basic residue arginine (R) to alanine (A) at position 42 disrupted M's nuclear localization. Moreover, a recombinant virus with R42A mutation in the M protein reduced viral replication in DF-1 cells and attenuated the virulence and pathogenicity of the virus in chickens. This is the first report to show that a basic residue mutation in the NDV M protein abrogates its nuclear localization and attenuates viral replication and pathogenicity.


Subject(s)
Mutation, Missense , Newcastle disease virus/physiology , Newcastle disease virus/pathogenicity , Point Mutation , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , Virus Replication , Amino Acid Substitution , Animal Structures/pathology , Animals , Cell Line , Cell Nucleus/chemistry , Chickens , Cytoplasm/chemistry , Disease Models, Animal , Histocytochemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Newcastle Disease/pathology , Newcastle Disease/virology , Newcastle disease virus/genetics , Protein Transport , Virulence
10.
Virology ; 452-453: 212-22, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24606698

ABSTRACT

The cellular nucleolar proteins are reported to facilitate the replication cycles of some human and animal viruses by interaction with viral proteins. In this study, a nucleolar phosphoprotein B23 was identified to interact with Newcastle disease virus (NDV) matrix (M) protein. We found that NDV M protein accumulated in the nucleolus by binding B23 early in infection, but resulted in the redistribution of B23 from the nucleoli to the nucleoplasm later in infection. In vitro binding studies utilizing deletion mutants indicated that amino acids 30-60 of M and amino acids 188-245 of B23 were required for binding. Furthermore, knockdown of B23 by siRNA or overexpression of B23 or M-binding B23-derived polypeptides remarkably reduced cytopathic effect and inhibited NDV replication. Collectively, we show that B23 facilitates NDV replication by targeting M to the nucleolus, demonstrating for the first time a direct role for nucleolar protein B23 in a paramyxovirus replication process.


Subject(s)
Cell Nucleolus/metabolism , Newcastle Disease/virology , Newcastle disease virus/physiology , Nuclear Proteins/metabolism , Poultry Diseases/metabolism , Viral Matrix Proteins/metabolism , Virus Replication , Animals , Cell Nucleolus/virology , Chickens , Newcastle Disease/metabolism , Newcastle disease virus/genetics , Nuclear Proteins/genetics , Nucleophosmin , Poultry Diseases/genetics , Poultry Diseases/virology , Protein Binding , Protein Transport , Viral Matrix Proteins/genetics
11.
Arch Virol ; 159(7): 1813-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24477785

ABSTRACT

The FPIV-like late domains identified in the matrix (M) proteins of parainfluenza virus 5 and mumps virus have been demonstrated to be critical for virus budding. In this study, we found that the same FPIV sequence motif is present in the N-terminus of the Newcastle disease virus (NDV) M protein. Mutagenesis experiments demonstrated that mutation of either phenylalanine (F) or proline (P) to alanine led to a more obvious decrease in viral virulence and replication and resulted in poor budding of the mutant viruses. Additionally, evidence for the involvement of cellular multivesicular body (MVB) proteins was obtained, since NDV production was inhibited upon expression of dominant-negative versions of the VPS4A-E228Q protein. Together, these results demonstrate that the FPIV motif, especially the residues F and P, within the NDV M protein, plays a critical role in NDV replication and budding, and this budding process likely involves the cellular MVB pathway.


Subject(s)
Newcastle disease virus/metabolism , Viral Matrix Proteins/metabolism , Virus Release/physiology , Virus Replication/physiology , Amino Acid Motifs , Mutation , Newcastle disease virus/genetics , Newcastle disease virus/physiology , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/genetics
12.
Arch Virol ; 159(2): 339-43, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23963518

ABSTRACT

CK10 and GS10 are two H5N1 highly pathogenic influenza viruses of similar genetic background but differ in their pathogenicity in mallard ducks. CK10 is highly pathogenic whereas GS10 is low pathogenic. In this study, strong inflammatory response in terms of the expression level of several cytokines was observed in mallard duck peripheral blood mononuclear cells (PBMC) infected with CK10 while mild response was triggered in those by GS10 infection. Two remarkable and intense peaks of immune response were induced by CK10 infection within 24 hours (at 8 and 24 hours post infection, respectively) without reducing the virus replication. Our observations indicated that sustained and intense innate immune responses may be central to the high pathogenicity caused by CK10 in ducks.


Subject(s)
Ducks/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza in Birds/immunology , Influenza in Birds/virology , Leukocytes, Mononuclear/immunology , Animals , Cytokines/biosynthesis , Gene Expression , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/physiology , Time Factors , Virus Replication
13.
Emerg Infect Dis ; 19(12): 2021-4, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24274396

ABSTRACT

We characterized 7 highly pathogenic avian influenza A(H5N1) viruses isolated from poultry in China during 2009-2012 and found that they belong to clade 2.3.4 but do not fit within the 3 defined subclades. Antigenic drift in subtype H5N1 variants may reduce the efficacy of vaccines designed to control these viruses in poultry.


Subject(s)
Genotype , Influenza A Virus, H5N1 Subtype/classification , Influenza A Virus, H5N1 Subtype/genetics , Influenza in Birds/epidemiology , Influenza in Birds/virology , Animals , Chickens , China/epidemiology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Phylogeny , Poultry/virology , Poultry Diseases/epidemiology , Poultry Diseases/virology
14.
Infect Genet Evol ; 20: 34-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23917026

ABSTRACT

Pasteurella multocida, the causative agent of fowl cholera, is a serious threat to poultry farming. In this study, we isolated and identified 40 P. multocida strains in fowl cholera outbreaks in Jiangsu province, China. The identified P. multocida was further characterized using multi-locus sequence typing (MLST). All of the 40 P. multocida strains studied are genetically identical and belong to the ST129 sequence type based on seven MLST loci. Our study provides evidence of a circulating epidemic strain of P. multocida in Jiangsu, China.


Subject(s)
Pasteurella Infections/veterinary , Pasteurella multocida/isolation & purification , Poultry Diseases/microbiology , Animals , China/epidemiology , DNA, Ribosomal/genetics , Disease Outbreaks , Genetic Variation , Genotype , Multilocus Sequence Typing , Pasteurella Infections/epidemiology , Pasteurella Infections/microbiology , Pasteurella multocida/classification , Pasteurella multocida/genetics , Poultry/microbiology , Poultry Diseases/epidemiology , RNA, Ribosomal, 16S/genetics , Serotyping
15.
J Virol ; 87(20): 11063-75, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23926340

ABSTRACT

Most highly pathogenic avian influenza A viruses cause only mild clinical signs in ducks, serving as an important natural reservoir of influenza A viruses. However, we isolated two H5N1 viruses that are genetically similar but differ greatly in virulence in ducks. A/Chicken/Jiangsu/k0402/2010 (CK10) is highly pathogenic, whereas A/Goose/Jiangsu/k0403/2010 (GS10) is low pathogenic. To determine the genetic basis for the high virulence of CK10 in ducks, we generated a series of single-gene reassortants between CK10 and GS10 and tested their virulence in ducks. Expression of the CK10 PA or hemagglutinin (HA) gene in the GS10 context resulted in increased virulence and virus replication. Conversely, inclusion of the GS10 PA or HA gene in the CK10 background attenuated the virulence and virus replication. Moreover, the PA gene had a greater contribution. We further determined that residues 101G and 237E in the PA gene contribute to the high virulence of CK10. Mutations at these two positions produced changes in virulence, virus replication, and polymerase activity of CK10 or GS10. Position 237 plays a greater role in determining these phenotypes. Moreover, the K237E mutation in the GS10 PA gene increased PA nuclear accumulation. Mutant GS10 viruses carrying the CK10 HA gene or the PA101G or PA237E mutation induced an enhanced innate immune response. A sustained innate response was detected in the brain rather than in the lung and spleen. Our results suggest that the PA and HA gene-mediated high virus replication and the intense innate immune response in the brain contribute to the high virulence of H5N1 virus in ducks.


Subject(s)
Brain/virology , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Immunity, Innate , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza in Birds/pathology , Viral Load , Virulence Factors/metabolism , Animals , Brain/immunology , DNA Mutational Analysis , Disease Models, Animal , Ducks , Genetic Engineering , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/isolation & purification , Influenza in Birds/virology , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Reassortant Viruses/genetics , Reassortant Viruses/immunology , Reassortant Viruses/isolation & purification , Reassortant Viruses/pathogenicity , Viral Proteins/genetics , Viral Proteins/metabolism , Virulence , Virulence Factors/genetics , Virus Replication
16.
J Virol ; 87(5): 2660-72, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23255810

ABSTRACT

Highly pathogenic H5N1 influenza A virus remains a substantial threat to public health. To understand the molecular basis and host mechanism for the high virulence of H5N1 viruses in mammals, we compared two H5N1 isolates which have similar genetic backgrounds but greatly differ in their virulence in mice. A/Chicken/Jiangsu/k0402/2010 (CK10) is highly pathogenic, whereas A/Goose/Jiangsu/k0403/2010 (GS10) is nonpathogenic. We first showed that CK10 elicited a more potent innate immune response than did GS10 in mouse lungs by increasing the number and expression levels of activated genes. We then generated a series of reassortants between the two viruses and evaluated their virulence in mice. Inclusion of the CK10 PA gene in the GS10 background resulted in a dramatic increase in virulence. Conversely, expression of the GS10 PA gene in the CK10 background significantly attenuated the virulence. These results demonstrated that the PA gene mainly determines the pathogenicity discrepancy between CK10 and GS10 in mice. We further determined that arginine (R) at position 353 of the PA gene contributes to the high virulence of CK10 in mice. The reciprocal substitution at position 353 in PA or the exchange of the entire PA gene largely caused the transfer of viral phenotypes, including virus replication, polymerase activity, and manipulation of the innate response, between CK10 and GS10. We therefore defined a novel molecular marker associated with the high virulence of H5N1 influenza viruses, providing further insights into the pathogenesis of H5N1 viruses in mammals.


Subject(s)
Immunity, Innate , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/pathogenicity , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , RNA-Dependent RNA Polymerase/genetics , Viral Proteins/genetics , Amino Acid Substitution , Animals , Chick Embryo , Female , Genes, Viral , HEK293 Cells , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Inflammation/immunology , Inflammation/virology , Influenza A Virus, H5N1 Subtype/immunology , Lung/immunology , Lung/virology , Mice , Mice, Inbred BALB C , Nucleocapsid Proteins , Orthomyxoviridae Infections/pathology , RNA-Binding Proteins/genetics , RNA-Dependent RNA Polymerase/chemistry , Reassortant Viruses/pathogenicity , Viral Core Proteins/genetics , Viral Proteins/chemistry , Virus Replication
17.
Bing Du Xue Bao ; 25(2): 117-24, 2009 Mar.
Article in Chinese | MEDLINE | ID: mdl-19678566

ABSTRACT

Twenty Newcastle disease virus (NDV) strains were isolated from chickens and geese in the field outbreaks during 2005 and 2006 in some regions of Jiangsu and Guangxi province. Assessment of the virulence by MDT and ICPI, RT-PCR and sequence analysis of fusion protein gene were used to compare the properties of NDV isolates. The results indicated that MDT and ICPI of the isolates were 45.3h - 58.2h and 1.61 - 2.00 respectively, which confirmed that the all NDV isolates were highly virulent. And their hemagglutinin were not resistant to heat and belonged to fast pattern of elution. The results of nucleotide sequencing and phylogentic analysis of fusion protein gene showed that the twenty strains shared homology from 79.7% to 100% among themselves, from 78.1% to 83.4% and from 80.2% to 90.1% with NDV LaSota, F48E8, respectively. The putative amino acid sequences of fusion protein at the cleavage sites of all the isolates were 112R-R-Q-R/K-R-F117, with the motif characteristics of the virulent NDV strain, which was in accordant with the results of assessment of the pathogenicity. The phylogentic tree based on sequences of fusion protein gene variable regions (47-420nt) revealed that the 18 strains belonged to sub-genotype VIId and the others belonged to an old genotype III of NDV, revealing that subgenotype VIId virus was responsible for the NDV outbreaks in some regions of Jiangsu and Guangxi promince recently.


Subject(s)
Disease Outbreaks , Newcastle disease virus/genetics , Viral Fusion Proteins/genetics , Amino Acid Motifs , Amino Acid Sequence , Animals , Base Sequence , Chickens/virology , China/epidemiology , Geese/virology , Molecular Epidemiology , Newcastle Disease/epidemiology , Newcastle Disease/genetics , Newcastle disease virus/pathogenicity , Phylogeny , Poultry Diseases/epidemiology , Poultry Diseases/genetics , Poultry Diseases/virology , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...